1
|
Li J, Wang H, Gao P, Wang R, Chan CL, Yi-Tsun Kao R, Li H, Sun H. Bismuth drug eradicates multi-drug resistant Burkholderia cepacia complex via aerobic respiration. Chem Sci 2025:d5sc02049b. [PMID: 40438165 PMCID: PMC12107623 DOI: 10.1039/d5sc02049b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 05/07/2025] [Indexed: 06/01/2025] Open
Abstract
Burkholderia cepacia complex (Bcc) is a group of Gram-negative opportunistic pathogens highly responsible for chronic pulmonary infection in cystic fibrosis (CF). Current therapies involving double or triple antibiotic combinations can rarely eradicate the pathogen in chronically infected patients owing to its intrinsic resistance to a variety of antibiotics. Herein, we show that a bismuth drug (and related compounds) could inhibit the growth of clinically antibiotic-resistant Bcc strains, with MIC (ca. 25 μg mL-1) comparable to that for Helicobacter pylori, and the combination of a bismuth drug and antibiotics also demonstrated excellent activity against biofilm and persisters of Bcc. Importantly, the in vitro antimicrobial activity of a bismuth drug could be well translated into in vivo evidenced by about 50% survival rates in the Galleria mellonella infection model. Transcriptomics analysis shows the dynamic responses of Bcc to bismuth treatment. Using a homemade metalloproteomic approach, we could identify 26 BiIII-binding proteins (15 cytosolic proteins and 11 membrane proteins). Further mechanistic studies reveal that bismuth drugs initially target the TCA cycle through the binding and inactivation of a series of enzymes including malate dehydrogenase (MDH), malate synthase (AceB), and succinyl coenzyme A synthetase (SCS), then interfere oxidative phosphorylation through binding to terminal oxidases, i.e., CyoC and CydA, to disrupt electron transport chain, eventually, disrupt protein translation and ribosome via binding and down-regulation of key proteins. Our studies highlight the great potential of bismuth drugs and/or compounds to treat multidrug-resistant Bcc infections.
Collapse
Affiliation(s)
- Jingru Li
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
- Chemistry and Chemical Engineering Guangdong Laboratory Shantou 515031 P. R. China
| | - Haibo Wang
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
| | - Peng Gao
- Applied Oral Sciences & Community Dental Care Division, Faculty of Dentistry, The University of Hong Kong Hong Kong SAR P. R. China
| | - Runming Wang
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
| | - Chun-Lung Chan
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
| | - Richard Yi-Tsun Kao
- Department of Microbiology, The University of Hong Kong Hong Kong SAR P. R. China
| | - Hongyan Li
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
| | - Hongzhe Sun
- Department of Chemistry, The University of Hong Kong Hong Kong SAR P. R. China
| |
Collapse
|
2
|
Wang C, Wei X, Zhong L, Chan CL, Li H, Sun H. Metal-Based Approaches for the Fight against Antimicrobial Resistance: Mechanisms, Opportunities, and Challenges. J Am Chem Soc 2025; 147:12361-12380. [PMID: 40063057 PMCID: PMC12007004 DOI: 10.1021/jacs.4c16035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/17/2025]
Abstract
The rapid emergency and spread of antimicrobial-resistant (AMR) bacteria and the lack of new antibiotics being developed pose serious threats to the global healthcare system. Therefore, the development of more effective therapies to overcome AMR is highly desirable. Metal ions have a long history of serving as antimicrobial agents, and metal-based compounds are now attracting more interest from scientific communities in the fight against AMR owing to their unique mechanism. Moreover, they may also serve as antibiotic adjuvants to enhance the efficacy of clinically used antibiotics. In this perspective, we highlight important showcase studies in the last 10 years on the development of metal-based strategies to overcome the AMR crisis. Specifically, we categorize these metallo-antimicrobials into five classes based on their modes of action (i.e., metallo-enzymes and metal-binding enzyme inhibitors, membrane perturbants, uptake/efflux system inhibitors/regulators, persisters inhibitors, and oxidative stress inducers). The significant advantages of metallo-antimicrobials over traditional antibiotics lie in their multitargeted mechanisms, which render less likelihood to generate resistance. However, we notice that such modes of action of metallo-antimicrobials may also raise concern over their potential side effects owing to the low selectivity toward pathogens and host, which appears to be the biggest obstacle for downstream translational research. We anticipate that combination therapy through repurposing (metallo)drugs with antibiotics and the optimization of their absorption route through formulation to achieve a target-oriented delivery will be a powerful way to combat AMR. Despite significant challenges, metallo-antimicrobials hold great opportunities for the therapeutic intervention of infection by resistant bacteria.
Collapse
Affiliation(s)
- Chenyuan Wang
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Xueying Wei
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- Department
of Microbiology, The University of Hong
Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Liang Zhong
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Chun-Lung Chan
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Hongyan Li
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- State
Key Laboratory of Synthetic Chemistry, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| | - Hongzhe Sun
- Department
of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- CAS-HKU
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
- State
Key Laboratory of Synthetic Chemistry, The
University of Hong Kong, Pokfulam Road, Hong Kong SAR, PR China
| |
Collapse
|
3
|
Singh I, Upadhyay A, Mandal AA, Saha S, Pragya P, Pradhan L, Nayak M, Dutta A, Agrawal AK, Mukherjee S, Banerjee S. Fe(II)-Photoantibiotics for Potential Antibacterial, Antibiofilm, and Infective Wound Healing Applications in Rat Model. J Med Chem 2025; 68:4453-4465. [PMID: 39913912 DOI: 10.1021/acs.jmedchem.4c02405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Herein, five Fe(II) complexes, viz., [Fe(N,N,N)2](ClO4)2, where N,N,N = 4'-phenyl-2,2':6',2″-terpyridine (Ph-tpy, Fe1), 4'-(4 aminophenyl) 2,2':6',2″ terpyridine (NH2-Phtpy, Fe2), 4-([2,2':6',2''-terpyridin]-4'-yl)-N,N-dimethylaniline (NMe2-Phtpy, Fe3), 4'-(p-nitrophenyl)-2.2':6',2″-terpyridine (NO2-Phtpy, Fe4), and 4'-(4-trifluoromethylphenyl)-2,2':6',2''-terpyridine (CF3-Phtpy, Fe5) were developed and screened for their visible-light-triggered antibacterial activity. Fe1-Fe5 exhibited absorption at ca. 450-600 nm, beneficial for antibacterial photodynamic therapy (aPDT) under visible light exposure. The excellent photostability and ideal energy gap between T1 and S0 of the complexes made them good photosensitizers for aPDT. Fe5 had the best antibacterial activity against Escherichia coli and Bacillus subtilis upon exposure to 400-700 nm (10 J cm-2) light due to reactive oxygen species (ROS) generation. Further, Fe5 showed antibiofilm activity on different medical-grade biomaterials and devices. Biocompatibility of Fe5 was validated using in vivo and chicken embryonic models (in ovo). Moreover, in vivo studies showed that Fe5 efficiently healed infected wounds within 9 days.
Collapse
Affiliation(s)
- Ishwar Singh
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Anjali Upadhyay
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Arif Ali Mandal
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sukanta Saha
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Pragya Pragya
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Lipi Pradhan
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Malay Nayak
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Arnab Dutta
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, Maharashtra 400076, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
4
|
Ip TKY, Wang Y, Wang S, Pu K, Wang R, Han B, Gao P, Xie Y, Kao RY, Ho PL, Li H, Sun H. Hinokitiol potentiates antimicrobial activity of bismuth drugs: a combination therapy for overcoming antimicrobial resistance. RSC Med Chem 2025:d4md00860j. [PMID: 40027343 PMCID: PMC11865920 DOI: 10.1039/d4md00860j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025] Open
Abstract
Antimicrobial resistance (AMR) poses a significant global health threat, rendering many infections untreatable. To combat AMR, repurposing approved drugs has emerged as a cost-effective strategy. Bismuth drugs, when combined with antibiotics, have been proven to be effective against Helicobacter pylori, including antibiotic-resistant strains. However, bismuth drugs alone exhibit limited antimicrobial activity against a narrow spectrum of pathogens. Therefore, a novel approach to enhance the efficacy and broaden the antimicrobial spectrum of bismuth drugs is highly desirable. Herein, we show that a naturally occurring monoterpenoid, hinokitiol, could potentiate the antimicrobial activity of bismuth drugs. We demonstrate a strong synergy between hinokitiol and colloidal bismuth subcitrate (CBS) against various Gram-positive and Gram-negative bacterial strains, including methicillin-resistant Staphylococcus aureus (MRSA). Moreover, the combination of hinokitiol and CBS exhibits anti-biofilm activity by preventing biofilm formation and eliminating S. aureus persister cells. Importantly, the combination therapy demonstrates promising antimicrobial efficacy in murine infection models including skin wound, gastrointestinal and blood infections. Mechanistic studies reveal that hinokitiol enhances bismuth ion (Bi(iii)) accumulation and reduces intracellular iron levels. By using thermal proteome profiling combined with dynamic quantitative proteomics analysis, we demonstrate that the bismuth-hinokitiol combination propagated the bismuth binding and interfered with ribosome synthesis, the glycolysis process, impaired bacterial cell wall synthesis and pathogenesis in MRSA. Our finding highlights the potential of combinatorial hinokitiol and bismuth drugs in the fight against AMR.
Collapse
Affiliation(s)
- Tiffany Ka-Yan Ip
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Yuchuan Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Suyu Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Keyuan Pu
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Runming Wang
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Bingjie Han
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Peng Gao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Yanxuan Xie
- School of Chemistry, Sun Yat-Sen University Guangzhou P. R. China
| | - Richard Y Kao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Pak-Leung Ho
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong Pokfulam Hong Kong SAR P. R. China
| | - Hongyan Li
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| | - Hongzhe Sun
- Department of Chemistry and HKU-CAS Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong Hong Kong SAR P. R. China
| |
Collapse
|
5
|
Esarev IV, Ott I. Modulation of the mechanism of action of antibacterial silver N-heterocyclic carbene complexes by variation of the halide ligand. RSC Adv 2025; 15:1782-1791. [PMID: 39835216 PMCID: PMC11744769 DOI: 10.1039/d4ra08093a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The antimicrobial properties of silver and silver complexes have been known in medicine since ancient times. However, limitations in stability and solubility have impaired medicinal chemistry and drug development research. With the advent of N-heterocyclic carbenes (NHC) as ligands, the development of synthesis methods for organometallic silver species of the type (NHC)AgX (where X = halide) has brought significant improvements, and the class of antimicrobial silver NHC complexes has emerged. However, reports studying structure-activity relationships and the mechanism of action of this compound type are still rare. This paper explores the activity of silver NHC complexes with halide (chloride, iodide) ligands and attempts to elucidate their mechanism of antibacterial action in Gram-negative E. coli bacteria in comparison to non-organometallic silver nitrate. In particular, the complexes with an iodide ligand were confirmed to cause stronger antibacterial effects in E. coli than silver nitrate. Moreover, iodide complexes exhibit an enhanced cellular uptake, show signs of DNA condensation, strongly inhibit TrxR in E. coli and cause a strong depolarization of the membrane potential and permeability of the inner cell membrane. In contrast, chloride silver NHC complexes and silver nitrate caused permeability of the outer membranes and also showed a different activity pattern in most of the studied mechanisms. In conclusion, by variation of the halide ligand of silver NHC complexes the mechanism of action and strength of antibacterial activity can be fine-tuned.
Collapse
Affiliation(s)
- Igor V Esarev
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstr. 55 38106 Braunschweig Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstr. 55 38106 Braunschweig Germany
| |
Collapse
|
6
|
Sahu P, Chakraborty S, Isab AA, Mandal SM, Dinda J. Biofilm Demolition by [Au III(N N)Cl(NHC)][PF 6] 2 Complexes Fastened with Bipyridine and Phenanthroline Ligands; Potent Antibacterial Agents Targeting Membrane Lipid. Chempluschem 2025; 90:e202400543. [PMID: 39434616 DOI: 10.1002/cplu.202400543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
The development of new antibacterial drugs is essential for staying ahead of evolving antibiotic resistant bacterial (ARB) threats, ensuring effective treatment options for bacterial infections, and protecting public health. Herein, we successfully designed and synthesized two novel gold(III)- NHC complexes, [Au(1)(bpy)Cl][PF6]2 (2) and [Au(1)(phen)Cl][PF6]2 (3) based on the proligand pyridyl[1,2-a]{2-pyridylimidazol}-3-ylidene hexafluorophosphate (1⋅HPF6) [bpy=2,2'-bipyridine; phen=1,10-phenanthroline]. The synthesized complexes were characterized spectroscopically; their geometries and structural arrangements were confirmed by single crystal XRD analysis. Complexes 2 and 3 showed photoluminescence properties at room temperature and the time-resolved fluorescence decay confirmed the fluorescence lifetimes of 0.54 and 0.62 ns respectively; which were used to demonstrate their direct interaction with bacterial cells. Among the two complexes, complex 3 was found to be more potent against the bacterial strains (Staphylococcus aureus, Gram-positive and Pseudomonas aeruginosa, Gram-negative bacteria) with the MIC values of 8.91 μM and 17.82 μM respectively. Studies revealed the binding of the complexes with the fundamental phospholipids present in the cell membrane of bacteria, which was found to be the leading cause of bacterial cell death. Cytotoxicity was evaluated using an MTT assay on 293 T cell lines; emphasizing the potential therapeutic uses of the Au(III)-NHC complexes to control bacterial infections.
Collapse
Affiliation(s)
- Priyanka Sahu
- Department of Chemistry, Utkal University, Vani Vihar, Bhubaneswar, Odisha, 751004, India
| | - Sourav Chakraborty
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - A A Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran, 31261, Saudi Arabia
| | - Santi M Mandal
- Department of Bioscience and Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Joydev Dinda
- Department of Chemistry, Utkal University, Vani Vihar, Bhubaneswar, Odisha, 751004, India
| |
Collapse
|
7
|
Castagliuolo G, Di Napoli M, Zangmo T, Szpunar J, Ronga L, Zanfardino A, Varcamonti M, Tesauro D. Antimicrobial Activity and Mode of Action of N-Heterocyclic Carbene Silver(I) Complexes. Molecules 2024; 30:76. [PMID: 39795133 PMCID: PMC11722542 DOI: 10.3390/molecules30010076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
Silver drugs have played a vital role in human healthcare for the treatment of infections for many centuries. Currently, due to antibiotic resistance, a potential scenario or the application of silver complexes may arise as substitutes for conventional antibiotics. In this perspective, N-heterocyclic carbene (NHC) ligands have been selected as carrier molecules for silver ions. In this study, we selected two mono NHC-silver halide complexes: bromo[1,3-diethyl-4,5-bis(4-methoxyphenyl)imidazol-2-ylidene]silver(I) (Ag4MC) and chloro[2-pyridin- N-(2-ethylacetylamido)-2-yl-2H-imidazol-2-ylidene]silver(I) (Ag5MC), and two cationic bis NHC silver complexes: bis[1,3-diethyl-4,5-bis(4-methoxyphenyl)imidazol-2-ylidene]silver(I) (Ag4BC) and bis[2-pyridin-N-(2-ethylacetylamido)-2-yl-2H-imidazol-2-ylidene]silver(I) (Ag5BC). The inhibitory properties of the four complexes were evaluated for their antimicrobial potential against a set of Gram (+) and Gram (-) bacterial strains and the fungus C. albicans. In addition, further investigations were made using fluorescence and scanning electron microscopy (SEM) in order to gain more insights into the mechanism of action. Some preliminary information on the Ag target was obtained by analyzing the cytosol of E. coli treated with Ag5MC by size-exclusion chromatography (SEC) coupled with inductively coupled plasma mass spectrometry (ICP-MS).
Collapse
Affiliation(s)
- Giusy Castagliuolo
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Michela Di Napoli
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Tshering Zangmo
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Joanna Szpunar
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Luisa Ronga
- IPREM (Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux), CNRS, E2S UPPA, Université de Pau et des Pays de l’Adour, CEDEX 9, 64012 Pau, France; (T.Z.); (J.S.); (L.R.)
| | - Anna Zanfardino
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Mario Varcamonti
- Department of Biology, University of Naples “Federico II”, Via Cynthia 26, 80126 Naples, Italy; (G.C.); (M.D.N.); (M.V.)
| | - Diego Tesauro
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples “Federico II”, Via Montesano 49, 80131 Naples, Italy;
| |
Collapse
|
8
|
Tiemuer A, Zhao H, Chen J, Li H, Sun H. Lighting Up and Identifying Metal-Binding Proteins in Cells. JACS AU 2024; 4:4628-4638. [PMID: 39735929 PMCID: PMC11672145 DOI: 10.1021/jacsau.4c00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024]
Abstract
Metal ions, either essential or therapeutic, play critical roles in life processes or in the treatment of diseases. Proteins and enzymes are involved in metal homeostasis and the action of metallodrugs. Imaging and identifying these metal-binding proteins will facilitate the elucidation of metal-mediated life processes. The emerging research field of metallomics and metalloproteomics has significantly advanced our understanding of metal homeostasis and the roles that metals play in biology and medicine. Fluorescence-based metalloproteomics offers the possibility of not only visualization but also identification of metal-binding proteins in living cells and tissues. Herein, we summarize different strategies of labeling and tracking of metal-binding proteins with the aid of fluorescent probes. We highlight several examples as showcases of how this fluorescence-based metalloproteomics approach could be utilized in metallobiology and chemical biology. In conclusion, we also discuss the advantages and limitations of fluorescence-based metalloproteomics approaches and point out future directions of metalloproteomics including development of more sensitive and selective fluorescence probes, integration with other omics approaches, as well as application of emerging advanced super-resolution imaging techniques that utilize fluorescent molecules or proteins. We aim to attract more scientists to engage in this exciting field.
Collapse
Affiliation(s)
- Aliya Tiemuer
- Department of Chemistry and HKU-CAS
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, P.R. China
| | - Hongyu Zhao
- Department of Chemistry and HKU-CAS
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, P.R. China
| | - Jingxin Chen
- Department of Chemistry and HKU-CAS
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, P.R. China
| | - Hongyan Li
- Department of Chemistry and HKU-CAS
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, P.R. China
| | - Hongzhe Sun
- Department of Chemistry and HKU-CAS
Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, P.R. China
| |
Collapse
|
9
|
Oheix E, Daou TJ, Pieuchot L. Antimicrobial zeolites and metal-organic frameworks. MATERIALS HORIZONS 2024; 11:6222-6256. [PMID: 39291597 DOI: 10.1039/d4mh00259h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The current surge in antibiotic resistance and the emergence of pandemics have created an urgent need for novel antimicrobial strategies. The controlled release of antimicrobial active principles remains the most viable strategy to date, and transition metal ions currently represent the main alternative to antibiotics. In this review, we explore the potential of two types of materials, zeolites and metal-organic frameworks (MOFs), for the controlled release of antimicrobial active principles, notably transition metal ions. These materials have unique crystalline microporous structures that act as reservoirs, enabling sustained bactericidal effects in various applications such as coatings, packaging, and medical devices. However, there are currently no convenient and standardised methods for evaluating their metal ion release and antimicrobial efficacy. This work discusses analytical techniques and the proposed mechanisms of action while highlighting recent advances in film, membrane, and coating technologies. By addressing the current limitations, microporous materials can revolutionise antimicrobial approaches, offering enhanced effectiveness and long-term sustainability.
Collapse
Affiliation(s)
- Emmanuel Oheix
- Institut de Science des Matériaux de Mulhouse (IS2M), Université de Haute Alsace (UHA), CNRS, UMR 7361, 3 bis rue Alfred Werner, F-68093 Mulhouse, France.
- Université de Strasbourg (UniStra), F-67000 Strasbourg, France
| | - T Jean Daou
- Aptar CSP Technologies, 9 rue du Sandholz, Niederbronn les Bains, France.
| | - Laurent Pieuchot
- Institut de Science des Matériaux de Mulhouse (IS2M), Université de Haute Alsace (UHA), CNRS, UMR 7361, 3 bis rue Alfred Werner, F-68093 Mulhouse, France.
- Université de Strasbourg (UniStra), F-67000 Strasbourg, France
| |
Collapse
|
10
|
Krychowiak-Maśnicka M, Wojciechowska WP, Bogaj K, Bielicka-Giełdoń A, Czechowska E, Ziąbka M, Narajczyk M, Kawiak A, Mazur T, Szafranek B, Królicka A. The Substantial Role of Cell and Nanoparticle Surface Properties in the Antibacterial Potential of Spherical Silver Nanoparticles. Nanotechnol Sci Appl 2024; 17:227-246. [PMID: 39659544 PMCID: PMC11630726 DOI: 10.2147/nsa.s489407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Purpose Although it is well known that the size, shape, and surface chemistry affect the biological potential of silver nanoparticles (AgNPs), the published studies that have considered the influence of AgNP surface on antibacterial activity have not provided conclusive results. This is the first study whose objective was to determine the significance of the surface net charge of AgNPs on their antibacterial potential, attraction to bacterial cells, and cell envelope disruption, considering differences in bacterial surface properties. Methods We evaluated five commercial AgNP colloids with identical size and shape but different surface ligands. We thoroughly characterized their physicochemical properties, including the zeta potential, hydrodynamic diameter, and polydispersity index, and determined the minimal inhibitory concentration (MIC) and minimal bactericidal concentration (MBC), along with silver absorption into bacterial cells. Moreover, we investigated structural changes in bacteria treated with AgNPs by using a crystal violet assay and electron microscopy. Results The zeta potential of AgNPs ranged from -47.6 to +68.5 mV, with a hydrodynamic diameter of 29-87 nm and a polydispersity index of 0.349-0.863. Bacterial susceptibility varied significantly (0.5 ≤ MIC ≤ 256 µg Ag/mL; 1 ≤ MBC ≤ 256 µg Ag/mL); we found the lowest susceptibility in bacteria with a cell wall or a polysaccharide capsule. The most active AgNPs (0.5 ≤ MIC ≤ 32 µg Ag/mL; 2 ≤ MBC ≤ 64 µg Ag/mL) had a moderate surface charge (-21.5 and +14.9 mV). The antibacterial potential was unrelated to ion dissolution or cell envelope disruption, and bacterial cells absorbed less of the most active AgNPs (1.75-7.65%). Conclusion Contrary to previous reports, we found that a moderate surface charge is crucial for the antibacterial activity of AgNPs, and that a significant attraction of the nanoparticle to the cell surface reduces the antibacterial potential of AgNPs. These findings challenge the existing views on AgNP antibacterial mechanisms and interactions with bacterial cells.
Collapse
Affiliation(s)
- Marta Krychowiak-Maśnicka
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Biologically Active Compounds, Gdansk, Poland
| | - Weronika Paulina Wojciechowska
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Biologically Active Compounds, Gdansk, Poland
| | - Karolina Bogaj
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Biologically Active Compounds, Gdansk, Poland
| | | | - Ewa Czechowska
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Experimental and Translational Immunology, Gdansk, Poland
| | - Magdalena Ziąbka
- AGH University of Krakow, Faculty of Materials Science and Ceramics, Department of Ceramics and Refractories, Krakow, Poland
| | - Magdalena Narajczyk
- University of Gdansk, Faculty of Biology, Bioimaging Laboratory, Gdansk, Poland
| | - Anna Kawiak
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Plant Protection and Biotechnology, Gdansk, Poland
| | - Tomasz Mazur
- AGH University of Krakow, Academic Centre for Materials and Nanotechnology, Krakow, Poland
| | | | - Aleksandra Królicka
- University of Gdansk, Intercollegiate Faculty of Biotechnology, Laboratory of Biologically Active Compounds, Gdansk, Poland
| |
Collapse
|
11
|
Lieu MD, Dang TKT, Nguyen TH. Green synthesized silver nanoparticles, a sustainable approach for fruit and vegetable preservation: An overview. Food Chem X 2024; 23:101664. [PMID: 39148528 PMCID: PMC11324848 DOI: 10.1016/j.fochx.2024.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/12/2024] [Accepted: 07/14/2024] [Indexed: 08/17/2024] Open
Abstract
Nanotechnology in which silver nanoparticles (AgNPs) have received more interest in fruits and vegetables (FaV) preservation due to their anti-microorganism properties. There are various approaches to synthesizing AgNPs, in which biological reduction, especially plant extraction containing bioactive compounds, is considered non-toxic, eco-friendly, and economically viable. AgNPs can be applied for FaV preservation by immersing or incorporating AgNPs into the edible coating or wrapper film. Depending on the type of coating and the kind of FaV, choosing the coating components is necessary to ensure the anti-microorganism ability and improve preservation efficiency. This review highlights green-synthesized AgNPs for preserving FaV. The study covered the materials employed in the green synthesis of AgNPs, their effectiveness against microorganisms, the influence of AgNPs on film structure, safety properties, and various preservation strategies. Using plant or bacterial-synthesized AgNPs in edible coatings offers a sustainable approach to enhance safety, edibility, environmental friendliness, and FaV quality during storage.
Collapse
Affiliation(s)
- My Dong Lieu
- Faculty of Food Science and Technology, Ho Chi Minh City University of Industry and Trade, 140 Le Trong Tan Street, Tay Thanh Ward, Tan Phu District, Ho Chi Minh City, Viet Nam
- Department of Biotechnology, Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City, Viet Nam
- Vietnam National University-Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc, Ho Chi Minh City, Viet Nam
| | - Thi Kim Thuy Dang
- Department of Plant Cell Technology, Institute of Tropical Biology, Vietnam Academy of Science and Technology, 9/621 Xa lo Ha Noi Street, Linh Trung Ward, Thu Duc City, Ho Chi Minh City, Viet Nam
| | - Thuy Huong Nguyen
- Department of Biotechnology, Faculty of Chemical Engineering, Ho Chi Minh City University of Technology (HCMUT), 268 Ly Thuong Kiet Street, District 10, Ho Chi Minh City, Viet Nam
- Vietnam National University-Ho Chi Minh City (VNU-HCM), Linh Trung Ward, Thu Duc, Ho Chi Minh City, Viet Nam
| |
Collapse
|
12
|
Cybulski O, Quintana C, Siek M, Grzybowski BA. Stirring-Controlled Synthesis of Ultrastable, Fluorescent Silver Nanoclusters. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400306. [PMID: 38934325 DOI: 10.1002/smll.202400306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/09/2024] [Indexed: 06/28/2024]
Abstract
This paper describes how macroscopic stirring of a reaction mixture can be used to produce nanostructures exhibiting properties not readily achievable via other protocols. In particular, it is shown that by simply adjusting the stirring rate, a standard glutathione-based method-to date, used to produce only marginally stable fluorescent silver nanoclusters, Ag NCs-can be boosted to yield nanoclusters retaining fluorescence for unprecedented periods of over 2 years. This enhancement derives not simply from increased homogenization of the reaction mixture but mainly from an appropriately timed delivery of oxygen from above the reaction mixture. In effect, oxygen serves as a reagent that dictates size, structure, stability, and functional properties of the growing nanoobjects.
Collapse
Affiliation(s)
- Olgierd Cybulski
- Center for Algorithmic and Robotized Synthesis (CARS), Institute for Basic Science (IBS), Ulsan, 44919, South Korea
| | - Cristóbal Quintana
- Center for Algorithmic and Robotized Synthesis (CARS), Institute for Basic Science (IBS), Ulsan, 44919, South Korea
| | - Marta Siek
- Center for Algorithmic and Robotized Synthesis (CARS), Institute for Basic Science (IBS), Ulsan, 44919, South Korea
| | - Bartosz A Grzybowski
- Center for Algorithmic and Robotized Synthesis (CARS), Institute for Basic Science (IBS), Ulsan, 44919, South Korea
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea
| |
Collapse
|
13
|
Do T, Vaculciakova S, Kluska K, Peris-Díaz MD, Priborsky J, Guran R, Krężel A, Adam V, Zitka O. Antioxidant-related enzymes and peptides as biomarkers of metallic nanoparticles (eco)toxicity in the aquatic environment. CHEMOSPHERE 2024; 364:142988. [PMID: 39103097 PMCID: PMC11422181 DOI: 10.1016/j.chemosphere.2024.142988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
Increased awareness of the impact of human activities on the environment has emerged in recent decades. One significant global environmental and human health issue is the development of materials that could potentially have negative effects. These materials can accumulate in the environment, infiltrate organisms, and move up the food chain, causing toxic effects at various levels. Therefore, it is crucial to assess materials comprising nano-scale particles due to the rapid expansion of nanotechnology. The aquatic environment, particularly vulnerable to waste pollution, demands attention. This review provides an overview of the behavior and fate of metallic nanoparticles (NPs) in the aquatic environment. It focuses on recent studies investigating the toxicity of different metallic NPs on aquatic organisms, with a specific emphasis on thiol-biomarkers of oxidative stress such as glutathione, thiol- and related-enzymes, and metallothionein. Additionally, the selection of suitable measurement methods for monitoring thiol-biomarkers in NPs' ecotoxicity assessments is discussed. The review also describes the analytical techniques employed for determining levels of oxidative stress biomarkers.
Collapse
Affiliation(s)
- Tomas Do
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Silvia Vaculciakova
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Katarzyna Kluska
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Manuel David Peris-Díaz
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Jan Priborsky
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Roman Guran
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic
| | - Ondrej Zitka
- Department of Chemistry and Biochemistry, Faculty of AgriSciences, Mendel University in Brno, Zemedelska 1, 613 00, Brno, Czech Republic.
| |
Collapse
|
14
|
Zhou Y, Li H, Tse E, Sun H. Metal-detection based techniques and their applications in metallobiology. Chem Sci 2024; 15:10264-10280. [PMID: 38994399 PMCID: PMC11234822 DOI: 10.1039/d4sc00108g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Metals are essential for human health and play a crucial role in numerous biological processes and pathways. Gaining a deeper insight into these biological events will facilitate novel strategies for disease prevention, early detection, and personalized treatment. In recent years, there has been significant progress in the development of metal-detection based techniques from single cell metallome and proteome profiling to multiplex imaging, which greatly enhance our comprehension of the intricate roles played by metals in complex biological systems. This perspective summarizes the recent progress in advanced metal-detection based techniques and highlights successful applications in elucidating the roles of metals in biology and medicine. Technologies including machine learning that couple with single-cell analysis such as mass cytometry and their application in metallobiology, cancer biology and immunology are also emphasized. Finally, we provide insights into future prospects and challenges involved in metal-detection based techniques, with the aim of inspiring further methodological advancements and applications that are accessible to chemists, biologists, and clinicians.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| | - Hongyan Li
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| | - Eric Tse
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| | - Hongzhe Sun
- Department of Chemistry, CAS-HKU Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong Pokfulam Road Hong Kong SAR P. R. China
| |
Collapse
|
15
|
Turner RJ. The good, the bad, and the ugly of metals as antimicrobials. Biometals 2024; 37:545-559. [PMID: 38112899 PMCID: PMC11101337 DOI: 10.1007/s10534-023-00565-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 11/18/2023] [Indexed: 12/21/2023]
Abstract
We are now moving into the antimicrobial resistance (AMR) era where more antibiotic resistant bacteria are now the majority, a problem brought on by both misuse and over use of antibiotics. Unfortunately, the antibiotic development pipeline dwindled away over the past decades as they are not very profitable compounds for companies to develop. Regardless researchers over the past decade have made strides to explore alternative options and out of this we see revisiting historical infection control agents such as toxic metals. From this we now see a field of research exploring the efficacy of metal ions and metal complexes as antimicrobials. Such antimicrobials are delivered in a variety of forms from metal salts, alloys, metal complexes, organometallic compounds, and metal based nanomaterials and gives us the broad term metalloantimicrobials. We now see many effective formulations applied for various applications using metals as antimicrobials that are effective against drug resistant strains. The purpose of the document here is to step aside and begin a conversation on the issues of use of such toxic metal compounds against microbes. This critical opinion mini-review in no way aims to be comprehensive. The goal here is to understand the benefits of metalloantimicrobials, but also to consider strongly the disadvantages of using metals, and what are the potential consequences of misuse and overuse. We need to be conscious of the issues, to see the entire system and affect through a OneHealth vision.
Collapse
Affiliation(s)
- Raymond J Turner
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, AB, Canada.
| |
Collapse
|
16
|
Korczak L, Majewski P, Iwaniuk D, Sacha P, Matulewicz M, Wieczorek P, Majewska P, Wieczorek A, Radziwon P, Tryniszewska E. Molecular mechanisms of tigecycline-resistance among Enterobacterales. Front Cell Infect Microbiol 2024; 14:1289396. [PMID: 38655285 PMCID: PMC11035753 DOI: 10.3389/fcimb.2024.1289396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/27/2024] [Indexed: 04/26/2024] Open
Abstract
The global emergence of antimicrobial resistance to multiple antibiotics has recently become a significant concern. Gram-negative bacteria, known for their ability to acquire mobile genetic elements such as plasmids, represent one of the most hazardous microorganisms. This phenomenon poses a serious threat to public health. Notably, the significance of tigecycline, a member of the antibiotic group glycylcyclines and derivative of tetracyclines has increased. Tigecycline is one of the last-resort antimicrobial drugs used to treat complicated infections caused by multidrug-resistant (MDR) bacteria, extensively drug-resistant (XDR) bacteria or even pan-drug-resistant (PDR) bacteria. The primary mechanisms of tigecycline resistance include efflux pumps' overexpression, tet genes and outer membrane porins. Efflux pumps are crucial in conferring multi-drug resistance by expelling antibiotics (such as tigecycline by direct expelling) and decreasing their concentration to sub-toxic levels. This review discusses the problem of tigecycline resistance, and provides important information for understanding the existing molecular mechanisms of tigecycline resistance in Enterobacterales. The emergence and spread of pathogens resistant to last-resort therapeutic options stands as a major global healthcare concern, especially when microorganisms are already resistant to carbapenems and/or colistin.
Collapse
Affiliation(s)
- Lukasz Korczak
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Majewski
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Dominika Iwaniuk
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Pawel Sacha
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | | | - Piotr Wieczorek
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | | | - Anna Wieczorek
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| | - Piotr Radziwon
- Regional Centre for Transfusion Medicine, Bialystok, Poland
| | - Elzbieta Tryniszewska
- Department of Microbiological Diagnostics and Infectious Immunology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
17
|
He J, Ma Y, Niu X, Pei J, Yan R, Xu F, Ma J, Ma X, Jia S, Ma W. Silver nanoparticles induce endothelial cytotoxicity through ROS-mediated mitochondria-lysosome damage and autophagy perturbation: The protective role of N-acetylcysteine. Toxicology 2024; 502:153734. [PMID: 38290605 DOI: 10.1016/j.tox.2024.153734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
Silver nanoparticles (AgNPs) are used increasingly often in the biomedical field, but their potential deleterious effects on the cardiovascular system remain to be elucidated. The primary aim of this study was to evaluate the toxic effects, and the underlying mechanisms of these effects, of AgNPs on human umbilical vein endothelial cells (HUVECs), as well as the protective role of N-acetylcysteine (NAC) against cytotoxicity induced by AgNPs. In this study, we found that exposure to AgNPs affects the morphology and function of endothelial cells which manifests as decreased cell proliferation, migration, and angiogenesis ability. Mechanistically, AgNPs can induce excessive cellular production of reactive oxygen species (ROS), leading to damage to cellular sub-organs such as mitochondria and lysosomes. More importantly, our data suggest that AgNPs causes autophagy defect, inhibits mitophagy, and finally activates the mitochondria-mediated apoptosis signaling pathway and evokes cell death. Interestingly, treatment with ROS scavenger-NAC can effectively suppress AgNP-induced endothelial damage.Our results indicate that ROS-mediated mitochondria-lysosome injury and autophagy dysfunction are potential factors of endothelial toxicity induced by AgNPs. This study may provide new evidence for the cardiovascular toxicity of AgNPs and serve as a reference for the safe use of nanoparticles(NPs) in the future.
Collapse
Affiliation(s)
- Jing He
- Department of Geriatric and Special Medicine, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China; School of Clinical Medicine, Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Yunyun Ma
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 75004, Republic of China; Heart Centre, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Xudong Niu
- Yinchuan Maternity and Child Care Hospital, Yinchuan 75004, Republic of China
| | - Jiansheng Pei
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Ru Yan
- Heart Centre, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Fangjing Xu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Jing Ma
- Department of Geriatric and Special Medicine, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Xiaojuan Ma
- Department of Geriatric and Special Medicine, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China
| | - Shaobin Jia
- Heart Centre, General Hospital of Ningxia Medical University, Yinchuan 75004, Republic of China.
| | - Wanrui Ma
- Department of Geriatrics, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan 523000, Republic of China.
| |
Collapse
|
18
|
Coverdale JPC, Polepalli S, Arruda MAZ, da Silva ABS, Stewart AJ, Blindauer CA. Recent Advances in Metalloproteomics. Biomolecules 2024; 14:104. [PMID: 38254704 PMCID: PMC10813065 DOI: 10.3390/biom14010104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Interactions between proteins and metal ions and their complexes are important in many areas of the life sciences, including physiology, medicine, and toxicology. Despite the involvement of essential elements in all major processes necessary for sustaining life, metalloproteomes remain ill-defined. This is not only owing to the complexity of metalloproteomes, but also to the non-covalent character of the complexes that most essential metals form, which complicates analysis. Similar issues may also be encountered for some toxic metals. The review discusses recently developed approaches and current challenges for the study of interactions involving entire (sub-)proteomes with such labile metal ions. In the second part, transition metals from the fourth and fifth periods are examined, most of which are xenobiotic and also tend to form more stable and/or inert complexes. A large research area in this respect concerns metallodrug-protein interactions. Particular attention is paid to separation approaches, as these need to be adapted to the reactivity of the metal under consideration.
Collapse
Affiliation(s)
- James P. C. Coverdale
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Edgbaston B15 2TT, UK;
| | | | - Marco A. Z. Arruda
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Ana B. Santos da Silva
- Institute of Chemistry, Department of Analytical Chemistry, Universidade Estadual de Campinas, Campinas 13083-970, Brazil; (M.A.Z.A.); (A.B.S.d.S.)
| | - Alan J. Stewart
- School of Medicine, University of St. Andrews, St Andrews KY16 9TF, UK
| | | |
Collapse
|
19
|
Wang Y, Yang X, Zhang S, Ai J, Wang J, Chen J, Zhao L, Wang W, You H. Comparative proteomics unveils the bacteriostatic mechanisms of Ga(III) on the regulation of metabolic pathways in Pseudomonas aeruginosa. J Proteomics 2023; 289:105011. [PMID: 37776994 DOI: 10.1016/j.jprot.2023.105011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Gallium has a long history as a chemotherapeutic agent. The mechanisms of action of Ga(III)-based anti-infectives are different from conventional antibiotics, which primarily result from the chemical similarities of Ga(III) with Fe(III) and substitution of gallium into iron-dependent biological pathways. However, more aspects of the molecular mechanisms of Ga(III) against human pathogens, especially the effects on bacterial metabolic processes, remain to be understood. Herein, by using conventional quantitative proteomics, we identified the protein changes of Pseudomonas aeruginosa (P. aeruginosa) in response to Ga(NO3)3 treatment. We show that Ga(III) exhibits bacteriostatic mode of action against P. aeruginosa through affecting the expressions of a number of key enzymes in the main metabolic pathways, including glycolysis, TCA cycle, amino acid metabolism, and protein and nucleic acid biosynthesis. In addition, decreased expressions of proteins associated with pathogenesis and virulence of P. aeruginosa were also identified. Moreover, the correlations between protein expressions and metabolome changes in P. aeruginosa upon Ga(III) treatment were identified and discussed. Our findings thus expand the understanding on the antimicrobial mechanisms of Ga(III) that shed light on enhanced therapeutic strategies. BIOLOGICAL SIGNIFICANCE: Mounting evidence suggest that the efficacy and resistance of clinical antibiotics are closely related to the metabolic homeostasis in bacterial pathogens. Ga(III)-based compounds have been repurposed as antibacterial therapeutic candidates against antibiotics resistant pathogens, and represent a safe and promising treatment for clinical human infections, while more thorough understandings of how bacteria respond to Ga(III) treatment are needed. In the present study, we provide evidences at the proteome level that indicate Ga(III)-induced metabolic perturbations in P. aeruginosa. We identified and discussed the interference of Ga(III) on the expressions and activities of enzymes in the main metabolic pathways in P. aeruginosa. In view of our previous report that the antimicrobial efficacy of Ga(III) could be modulated according to Ga(III)-induced metabolome changes in P. aeruginosa, our current analyses may provide theoretical basis at the proteome level for the development of efficient gallium-based therapies by exploiting bacterial metabolic mechanisms.
Collapse
Affiliation(s)
- Yuchuan Wang
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China.
| | - Xue Yang
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Shuo Zhang
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Jiayi Ai
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Junteng Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Junxin Chen
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Lin Zhao
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Wanying Wang
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| | - Haoxin You
- Hebei Key Laboratory for Chronic Diseases, School of Basic Medical Sciences, China
| |
Collapse
|
20
|
Ma M, Zhao M, Ji R, Guo Y, Li D, Zeng S. Adjusting the Dose of Ag-Ion Implantation on TiN-Ag-Modified SLA-Ti Creates Different Micronanostructures: Implications on Bacteriostasis, Biocompatibility, and Osteogenesis in Dental Implants. ACS OMEGA 2023; 8:39269-39278. [PMID: 37901550 PMCID: PMC10601048 DOI: 10.1021/acsomega.3c04769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023]
Abstract
The prevention of aseptic loosening and peri-implantitis is crucial for the success of dental implant surgery. In this study, different doses of Ag-implanted TiN/Ag nanomultilayers were prepared on the sandblasting with large grit and acid etching (SLA)-Ti surface using a multiarc ion-plating system and an ion-implantation system, respectively. The physical and chemical properties of the samples were assessed using various techniques, including scanning electron microscopy, energy-dispersive spectroscopy, X-ray diffraction, X-ray photoelectron spectroscopy, atomic force microscopy, inductively coupled plasma atomic emission spectrometry, and water contact angle measurements. In addition, the applicability and biosafety of the SLA/1 × 1017-Ag and SLA/1 × 1018-Ag surfaces were determined via biocompatibility testing in vivo and in vitro. The results demonstrated that the physical and chemical properties of SLA/1 × 1017-Ag and SLA/1 × 1018-Ag surfaces were different to some extent. However, compared with SLA-Ti, silver-loaded TiN/Ag-modified SLA-Ti surfaces (SLA/1 × 1018-Ag) with enhanced bacteriostatis, osteogenesis, and biocompatibility have great potential for dental applications.
Collapse
Affiliation(s)
- Ming Ma
- Department
of Pediatric dentistry, School and Hospital of Stomatology, Guangdong
Engineering Research Center of Oral Restoration and Reconstruction,
Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative
Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Mengli Zhao
- School
of Electronic Engineering, Chaohu University, Anhui 238024, China
| | - Ruotong Ji
- Department
of Pediatric dentistry, School and Hospital of Stomatology, Guangdong
Engineering Research Center of Oral Restoration and Reconstruction,
Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative
Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Yi Guo
- Department
of Pediatric dentistry, School and Hospital of Stomatology, Guangdong
Engineering Research Center of Oral Restoration and Reconstruction,
Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative
Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| | - Dejun Li
- College
of Physics and Materials Science, Tianjin
Normal University, Tianjin 300387, China
| | - Sujuan Zeng
- Department
of Pediatric dentistry, School and Hospital of Stomatology, Guangdong
Engineering Research Center of Oral Restoration and Reconstruction,
Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative
Medicine, Guangzhou Medical University, Guangzhou, Guangdong 510182, China
| |
Collapse
|
21
|
Wu W, Duan M, Shao S, Meng F, Qin Y, Zhang M. Recent advances in nanomaterial-mediated bacterial molecular action and their applications in wound therapy. Biomater Sci 2023; 11:6748-6769. [PMID: 37665317 DOI: 10.1039/d3bm00663h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Because of the multi-pathway antibacterial mechanisms of nanomaterials, they have received widespread attention in wound therapy. However, owing to the complexities of bacterial responses toward nanomaterials, antibacterial molecular mechanisms remain unclear, making it difficult to rationally design highly efficient antibacterial nanomaterials. Fortunately, molecular dynamics simulations and omics techniques have been used as effective methods to further investigate the action targets of nanomaterials. Therefore, the review comprehensively analyzes the antibacterial mechanisms of nanomaterials from the morphology-dependent antibacterial activity and physicochemical/optical properties-dependent antibacterial activity, which provided guidance for constructing excellently efficient and broad-spectrum antibacterial nanomaterials for wound therapy. More importantly, the main molecular action targets of nanomaterials from the membranes, DNA, energy metabolism pathways, oxidative stress defense systems, ribosomes, and biofilms are elaborated in detail. Furthermore, nanomaterials used in wound therapy are reviewed and discussed. Finally, future directions of nanomaterials from mechanisms to nanomedicine are further proposed.
Collapse
Affiliation(s)
- Wanfeng Wu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| | - Mengjiao Duan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| | - Shuxuan Shao
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| | - Yanan Qin
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
22
|
Waters JE, Stevens-Cullinane L, Siebenmann L, Hess J. Recent advances in the development of metal complexes as antibacterial agents with metal-specific modes of action. Curr Opin Microbiol 2023; 75:102347. [PMID: 37467616 DOI: 10.1016/j.mib.2023.102347] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 07/21/2023]
Abstract
The mounting burden of antimicrobial resistance (AMR) is one of the most concerning threats to public health worldwide. With low economic incentives and a dwindling supply of new drugs in clinical pipelines, more innovative approaches to novel drug design and development are desperately required. Metal-based compounds are rapidly emerging as an alternative to organic drugs, as they have the ability to kill pathogens via metal-specific modes of action. We herein review recent advances in metal-based antibacterial agents, including metal complexes, metal ions and catalytic metallodrugs. The review concludes with a perspective on the rational design of metal-based antibiotics, and how we can exploit their unique properties to tackle AMR.
Collapse
Affiliation(s)
- Jessica E Waters
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lars Stevens-Cullinane
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Lukas Siebenmann
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom
| | - Jeannine Hess
- Biological Inorganic Chemistry Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom; Department of Chemistry, King's College London, Britannia House, 7 Trinity Street, London SE1 1DB, United Kingdom.
| |
Collapse
|
23
|
Korotkov SM. Mitochondrial Oxidative Stress Is the General Reason for Apoptosis Induced by Different-Valence Heavy Metals in Cells and Mitochondria. Int J Mol Sci 2023; 24:14459. [PMID: 37833908 PMCID: PMC10572412 DOI: 10.3390/ijms241914459] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/15/2023] Open
Abstract
This review analyzes the causes and consequences of apoptosis resulting from oxidative stress that occurs in mitochondria and cells exposed to the toxic effects of different-valence heavy metals (Ag+, Tl+, Hg2+, Cd2+, Pb2+, Al3+, Ga3+, In3+, As3+, Sb3+, Cr6+, and U6+). The problems of the relationship between the integration of these toxic metals into molecular mechanisms with the subsequent development of pathophysiological processes and the appearance of diseases caused by the accumulation of these metals in the body are also addressed in this review. Such apoptosis is characterized by a reduction in cell viability, the activation of caspase-3 and caspase-9, the expression of pro-apoptotic genes (Bax and Bcl-2), and the activation of protein kinases (ERK, JNK, p53, and p38) by mitogens. Moreover, the oxidative stress manifests as the mitochondrial permeability transition pore (MPTP) opening, mitochondrial swelling, an increase in the production of reactive oxygen species (ROS) and H2O2, lipid peroxidation, cytochrome c release, a decline in the inner mitochondrial membrane potential (ΔΨmito), a decrease in ATP synthesis, and reduced glutathione and oxygen consumption as well as cytoplasm and matrix calcium overload due to Ca2+ release from the endoplasmic reticulum (ER). The apoptosis and respiratory dysfunction induced by these metals are discussed regarding their interaction with cellular and mitochondrial thiol groups and Fe2+ metabolism disturbance. Similarities and differences in the toxic effects of Tl+ from those of other heavy metals under review are discussed. Similarities may be due to the increase in the cytoplasmic calcium concentration induced by Tl+ and these metals. One difference discussed is the failure to decrease Tl+ toxicity through metallothionein-dependent mechanisms. Another difference could be the decrease in reduced glutathione in the matrix due to the reversible oxidation of Tl+ to Tl3+ near the centers of ROS generation in the respiratory chain. The latter may explain why thallium toxicity to humans turned out to be higher than the toxicity of mercury, lead, cadmium, copper, and zinc.
Collapse
Affiliation(s)
- Sergey M Korotkov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Thorez pr. 44, 194223 St. Petersburg, Russia
| |
Collapse
|
24
|
Norton AE, Ewing R, Tilley M, Whitworth J, Cohnstaedt LW. Fatal Food: Silver-Coated Grain Particles Display Larvicidal Activity in Culex quinquefasciatus. ACS OMEGA 2023; 8:33437-33443. [PMID: 37744788 PMCID: PMC10515342 DOI: 10.1021/acsomega.3c03210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023]
Abstract
Mosquitoes pose a significant risk to millions of people worldwide since they can transmit pathogens. Current methods to control mosquito populations include the use of synthetic pesticides. Nanotechnology may be a solution to develop new mosquito control. However, one barrier to expanding the impact of nanomaterials is the ability to mass-produce the particles. Here, we report a novel hybrid particle synthesis combining micro- and nanoparticles using the coprecipitation technique with the potential for mass production. These particles may have applications as a mosquito larvacide. The particles reported here were designed using a microparticle zein polymer as the core and a nanoparticle silver as the active ingredient. The hybrid NPs reported here targeted a late-stage mosquito larvae and that resulted in a high larval mortality concentration (1.0 ppm, LC90) and suppression of pupal emergence at 0.1 ppm. This research demonstrates the efficacy of a plant-based core with a metal-based AI coating (AgNPs) against larval mosquitoes.
Collapse
Affiliation(s)
- Amie E. Norton
- Department
of Entomology, Kansas State University, Manhattan, Kansas 66506-0100, United
States
| | - Robert Ewing
- National
Bio and Agro-Defense Facility-USDA, Manhattan, Kansas 66503, United States
| | - Michael Tilley
- USDA,
Agricultural Research Service, Center for Grain and Animal Health
Research, 1515 College
Avenue, Manhattan, Kansas 66502, United States
| | - Jeff Whitworth
- Department
of Entomology, Kansas State University, Manhattan, Kansas 66506-0100, United
States
| | - Lee W. Cohnstaedt
- National
Bio and Agro-Defense Facility-USDA, Manhattan, Kansas 66503, United States
| |
Collapse
|
25
|
Pormohammad A, Firrincieli A, Salazar-Alemán DA, Mohammadi M, Hansen D, Cappelletti M, Zannoni D, Zarei M, Turner RJ. Insights into the Synergistic Antibacterial Activity of Silver Nitrate with Potassium Tellurite against Pseudomonas aeruginosa. Microbiol Spectr 2023; 11:e0062823. [PMID: 37409940 PMCID: PMC10433965 DOI: 10.1128/spectrum.00628-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
The constant, ever-increasing antibiotic resistance crisis leads to the announcement of "urgent, novel antibiotics needed" by the World Health Organization. Our previous works showed a promising synergistic antibacterial activity of silver nitrate with potassium tellurite out of thousands of other metal/metalloid-based antibacterial combinations. The silver-tellurite combined treatment not only is more effective than common antibiotics but also prevents bacterial recovery, decreases the risk of future resistance chance, and decreases the effective concentrations. We demonstrate that the silver-tellurite combination is effective against clinical isolates. Further, this study was conducted to address knowledge gaps in the available data on the antibacterial mechanism of both silver and tellurite, as well as to give insight into how the mixture provides synergism as a combination. Here, we defined the differentially expressed gene profile of Pseudomonas aeruginosa under silver, tellurite, and silver-tellurite combination stress using an RNA sequencing approach to examine the global transcriptional changes in the challenged cultures grown in simulated wound fluid. The study was complemented with metabolomics and biochemistry assays. Both metal ions mainly affected four cellular processes, including sulfur homeostasis, reactive oxygen species response, energy pathways, and the bacterial cell membrane (for silver). Using a Caenorhabditis elegans animal model we showed silver-tellurite has reduced toxicity over individual metal/metalloid salts and provides increased antioxidant properties to the host. This work demonstrates that the addition of tellurite would improve the efficacy of silver in biomedical applications. IMPORTANCE Metals and/or metalloids could represent antimicrobial alternatives for industrial and clinical applications (e.g., surface coatings, livestock, and topical infection control) because of their great properties, such as good stability and long half-life. Silver is the most common antimicrobial metal, but resistance prevalence is high, and it can be toxic to the host above a certain concentration. We found that a silver-tellurite composition has antibacterial synergistic effect and that the combination is beneficial to the host. So, the efficacy and application of silver could increase by adding tellurite in the recommended concentration(s). We used different methods to evaluate the mechanism for how this combination can be so incredibly synergistic, leading to efficacy against antibiotic- and silver-resistant isolates. Our two main findings are that (i) both silver and tellurite mostly target the same pathways and (ii) the coapplication of silver with tellurite tends not to target new pathways but targets the same pathways with an amplified change.
Collapse
Affiliation(s)
- Ali Pormohammad
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
- CCrest Laboratories, Inc., Montreal, Quebec, Canada
| | - Andrea Firrincieli
- Department for Innovation in Biological, Agro-Food and Forest systems, University of Tuscia, Viterbo, Italy
| | - Daniel A. Salazar-Alemán
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Mehdi Mohammadi
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Dave Hansen
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Martina Cappelletti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Davide Zannoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mohammad Zarei
- Renal Division, Brigham & Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- John B. Little Center for Radiation Sciences, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Raymond J. Turner
- Department of Biological Sciences, Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
26
|
O’Shaughnessy M, Sheils O, Baird AM. The Lung Microbiome in COPD and Lung Cancer: Exploring the Potential of Metal-Based Drugs. Int J Mol Sci 2023; 24:12296. [PMID: 37569672 PMCID: PMC10419288 DOI: 10.3390/ijms241512296] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer 17 are two of the most prevalent and debilitating respiratory diseases worldwide, both associated with high morbidity and mortality rates. As major global health concerns, they impose a substantial burden on patients, healthcare systems, and society at large. Despite their distinct aetiologies, lung cancer and COPD share common risk factors, clinical features, and pathological pathways, which have spurred increasing research interest in their co-occurrence. One area of particular interest is the role of the lung microbiome in the development and progression of these diseases, including the transition from COPD to lung cancer. Exploring novel therapeutic strategies, such as metal-based drugs, offers a potential avenue for targeting the microbiome in these diseases to improve patient outcomes. This review aims to provide an overview of the current understanding of the lung microbiome, with a particular emphasis on COPD and lung cancer, and to discuss the potential of metal-based drugs as a therapeutic strategy for these conditions, specifically concerning targeting the microbiome.
Collapse
Affiliation(s)
- Megan O’Shaughnessy
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| | - Orla Sheils
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, St. James’s Hospital, D08 RX0X Dublin, Ireland
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, D08 W9RT Dublin, Ireland
| |
Collapse
|
27
|
Ronga L, Varcamonti M, Tesauro D. Structure-Activity Relationships in NHC-Silver Complexes as Antimicrobial Agents. Molecules 2023; 28:molecules28114435. [PMID: 37298911 DOI: 10.3390/molecules28114435] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Silver has a long history of antimicrobial activity and received an increasing interest in last decades owing to the rise in antimicrobial resistance. The major drawback is the limited duration of its antimicrobial activity. The broad-spectrum silver containing antimicrobial agents are well represented by N-heterocyclic carbenes (NHCs) silver complexes. Due to their stability, this class of complexes can release the active Ag+ cations in prolonged time. Moreover, the properties of NHC can be tuned introducing alkyl moieties on N-heterocycle to provide a range of versatile structures with different stability and lipophilicity. This review presents designed Ag complexes and their biological activity against Gram-positive, Gram-negative bacteria and fungal strains. In particular, the structure-activity relationships underlining the major requirements to increase the capability to induce microorganism death are highlighted here. Moreover, some examples of encapsulation of silver-NHC complexes in polymer-based supramolecular aggregates are reported. The targeted delivery of silver complexes to the infected sites will be the most promising goal for the future.
Collapse
Affiliation(s)
- Luisa Ronga
- Institut des Sciences Analytiques et de Physico-Chimie Pour l'Environnement et les Matériaux, Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM, 64053 Pau, France
| | - Mario Varcamonti
- Department of Biology, University of Naples "Federico II", Via Cynthia, 80143 Naples, Italy
| | - Diego Tesauro
- Department of Pharmacy and Interuniversity Research Centre on Bioactive Peptides (CIRPeB), University of Naples "Federico II", Via Montesano, 49, 80131 Naples, Italy
| |
Collapse
|
28
|
Tan W, Tian Y, Zhang Q, Miao S, Wu W, Miao X, Kuang H, Yang W. Antioxidant and antibacterial activity of Apis laboriosa honey against Salmonella enterica serovar Typhimurium. Front Nutr 2023; 10:1181492. [PMID: 37252242 PMCID: PMC10211265 DOI: 10.3389/fnut.2023.1181492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a common food-borne pathogen that commonly causes gastroenteritis in humans and animals. Apis laboriosa honey (ALH) harvested in China has significant antibacterial activity against Staphylococcus aureus, Escherichia coli, and Bacillus subtilis. We hypothesize that ALH has antibacterial activity against S. Typhimurium. The physicochemical parameters, minimum inhibitory and bactericidal concentrations (MIC and MBC) and the possible mechanism were determined. The results showed that there were significantly different physicochemical parameters, including 73 phenolic compounds, among ALH samples harvested at different times and from different regions. Their antioxidant activity was affected by their components, especially total phenol and flavonoid contents (TPC, TFC), which had a high correlation with antioxidant activities except for the O2- assay. The MIC and MBC of ALH against S. Typhimurium were 20-30% and 25-40%, respectively, which were close to those of UMF5+ manuka honey. The proteomic experiment revealed the possible antibacterial mechanism of ALH1 at IC50 (2.97%, w/v), whose antioxidant activity reduced the bacterial reduction reaction and energy supply, mainly by inhibiting the citrate cycle (TCA cycle), amino acid metabolism pathways and enhancing the glycolysis pathway. The results provide a theoretical basis for the development of bacteriostatic agents and application of ALH.
Collapse
Affiliation(s)
- Weihua Tan
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Yuanyuan Tian
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
| | - Qingya Zhang
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Siwei Miao
- M.X.’s Expert Workstation, Pu’er, Yunnan, China
| | - Wenrong Wu
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
| | - Xiaoqing Miao
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
- M.X.’s Expert Workstation, Pu’er, Yunnan, China
| | - Haiou Kuang
- M.X.’s Expert Workstation, Pu’er, Yunnan, China
- Research Institute of Eastern Honeybee, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Wenchao Yang
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
- Bee Product Processing and Application Research Center of the Ministry of Education, Fuzhou, Fujian, China
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian, China
- M.X.’s Expert Workstation, Pu’er, Yunnan, China
| |
Collapse
|
29
|
Wang X, Zhang M, Zhu T, Wei Q, Liu G, Ding J. Flourishing Antibacterial Strategies for Osteomyelitis Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206154. [PMID: 36717275 PMCID: PMC10104653 DOI: 10.1002/advs.202206154] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/05/2022] [Indexed: 06/18/2023]
Abstract
Osteomyelitis is a destructive disease of bone tissue caused by infection with pathogenic microorganisms. Because of the complex and long-term abnormal conditions, osteomyelitis is one of the refractory diseases in orthopedics. Currently, anti-infective therapy is the primary modality for osteomyelitis therapy in addition to thorough surgical debridement. However, bacterial resistance has gradually reduced the benefits of traditional antibiotics, and the development of advanced antibacterial agents has received growing attention. This review introduces the main targets of antibacterial agents for treating osteomyelitis, including bacterial cell wall, cell membrane, intracellular macromolecules, and bacterial energy metabolism, focuses on their mechanisms, and predicts prospects for clinical applications.
Collapse
Affiliation(s)
- Xukai Wang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Mingran Zhang
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Tongtong Zhu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Qiuhua Wei
- Department of Disinfection and Infection ControlChinese PLA Center for Disease Control and Prevention20 Dongda StreetBeijing100071P. R. China
| | - Guangyao Liu
- Department of Thoracic SurgeryChina‐Japan Union Hospital of Jilin University126 Xiantai StreetChangchun130033P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
30
|
Skos L, Borutzki Y, Gerner C, Meier-Menches SM. Methods to identify protein targets of metal-based drugs. Curr Opin Chem Biol 2023; 73:102257. [PMID: 36599256 DOI: 10.1016/j.cbpa.2022.102257] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 01/03/2023]
Abstract
Metal-based anticancer agents occupy a distinct chemical space due to their particular coordination geometry and reactivity. Despite the initial DNA-targeting paradigm for this class of compounds, it is now clear that they can also be tuned to target proteins in cells, depending on the metal and ligand scaffold. Since metallodrug discovery is dominated by phenotypic screenings, tailored proteomics strategies were crucial to identify and validate protein targets of several investigative and clinically advanced metal-based drugs. Here, such experimental approaches are discussed, which showed that metallodrugs based on ruthenium, gold, rhenium and even platinum, can selectively and specifically target proteins with clear-cut down-stream effects. Target identification strategies are expected to support significantly the mechanism-driven clinical translation of metal-based drugs.
Collapse
Affiliation(s)
- Lukas Skos
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Yasmin Borutzki
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Doctoral School of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria
| | - Samuel M Meier-Menches
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria; Joint Metabolome Facility, University of Vienna and Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
31
|
Wang H, Hu L, Li H, Lai YT, Wei X, Xu X, Cao Z, Cao H, Wan Q, Chang YY, Xu A, Zhou Q, Jiang G, He ML, Sun H. Mitochondrial ATP synthase as a direct molecular target of chromium(III) to ameliorate hyperglycaemia stress. Nat Commun 2023; 14:1738. [PMID: 36977671 PMCID: PMC10050403 DOI: 10.1038/s41467-023-37351-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Chromium(III) is extensively used as a supplement for muscle development and the treatment of diabetes mellitus. However, its mode of action, essentiality, and physiological/pharmacological effects have been a subject of scientific debate for over half a century owing to the failure in identifying the molecular targets of Cr(III). Herein, by integrating fluorescence imaging with a proteomic approach, we visualized the Cr(III) proteome being mainly localized in the mitochondria, and subsequently identified and validated eight Cr(III)-binding proteins, which are predominately associated with ATP synthesis. We show that Cr(III) binds to ATP synthase at its beta subunit via the catalytic residues of Thr213/Glu242 and the nucleotide in the active site. Such a binding suppresses ATP synthase activity, leading to the activation of AMPK, improving glucose metabolism, and rescuing mitochondria from hyperglycaemia-induced fragmentation. The mode of action of Cr(III) in cells also holds true in type II diabetic male mice. Through this study, we resolve the long-standing question of how Cr(III) ameliorates hyperglycaemia stress at the molecular level, opening a new horizon for further exploration of the pharmacological effects of Cr(III).
Collapse
Affiliation(s)
- Haibo Wang
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Ligang Hu
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
| | - Hongyan Li
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Yau-Tsz Lai
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Xueying Wei
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Xiaohan Xu
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Zhenkun Cao
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Huiming Cao
- Institute of Environment and Health, Jianghan University, Wuhan, 430056, P.R. China
| | - Qianya Wan
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong, P.R. China
| | - Yuen-Yan Chang
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China
| | - Aimin Xu
- Department of Pharmacology and Pharmacy, and State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pok Fu Lam, Hong Kong, P.R. China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
| | - Ming-Liang He
- Department of Biomedical Science, City University of Hong Kong, Kowloon Tong, Hong Kong, P.R. China
| | - Hongzhe Sun
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, CAS-HKU Joint Laboratory of Metallomics on Health and Environment, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R., P.R. China.
| |
Collapse
|
32
|
Abstract
Bacteria, similar to most organisms, have a love-hate relationship with metals: a specific metal may be essential for survival yet toxic in certain forms and concentrations. Metal ions have a long history of antimicrobial activity and have received increasing attention in recent years owing to the rise of antimicrobial resistance. The search for antibacterial agents now encompasses metal ions, nanoparticles and metal complexes with antimicrobial activity ('metalloantibiotics'). Although yet to be advanced to the clinic, metalloantibiotics are a vast and underexplored group of compounds that could lead to a much-needed new class of antibiotics. This Review summarizes recent developments in this growing field, focusing on advances in the development of metalloantibiotics, in particular, those for which the mechanism of action has been investigated. We also provide an overview of alternative uses of metal complexes to combat bacterial infections, including antimicrobial photodynamic therapy and radionuclide diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Angelo Frei
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| | - Anthony D Verderosa
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Alysha G Elliott
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Johannes Zuegg
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Mark A T Blaskovich
- Community for Open Antimicrobial Drug Discovery, Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
33
|
Dixit H, Upadhyay V, Kulharia M, Verma SK. The putative metal-binding proteome of the Coronaviridae family. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2023; 15:6969429. [PMID: 36610727 DOI: 10.1093/mtomcs/mfad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023]
Abstract
Metalloproteins are well-known for playing various physicochemical processes in all life forms, including viruses. Some life-threatening viruses (such as some members of the Coronaviridae family of viruses) are emerged and remerged frequently and are rapidly transmitted throughout the globe. This study aims to identify and characterize the metal-binding proteins (MBPs) of the Coronaviridae family of viruses and further provides insight into the MBP's role in sustaining and propagating viruses inside a host cell and in the outer environment. In this study, the available proteome of the Coronaviridae family was exploited. Identified potential MBPs were analyzed for their functional domains, structural aspects, and subcellular localization. We also demonstrate phylogenetic aspects of all predicted MBPs among other Coronaviridae family members to understand the evolutionary trend among their respective hosts. A total of 256 proteins from 51 different species of coronaviruses are predicted as MBPs. These MBPs perform various key roles in the replication and survival of viruses within the host cell. Cysteine, aspartic acid, threonine, and glutamine are key amino acid residues interacting with respective metal ions. Our observations also indicate that the metalloproteins of this family of viruses circulated and evolved in different hosts, which supports the zoonotic nature of coronaviruses. The comprehensive information on MBPs of the Coronaviridae family may be further helpful in designing novel therapeutic metalloprotein targets. Moreover, the study of viral MBPs can also help to understand the roles of MBPs in virus pathogenesis and virus-host interactions.
Collapse
Affiliation(s)
- Himisha Dixit
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra176206, India
| | - Vipin Upadhyay
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra176206, India
| | - Mahesh Kulharia
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra176206, India
| | - Shailender Kumar Verma
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra176206, India.,Department of Environmental Studies, University of Delhi, Delhi110007, India
| |
Collapse
|
34
|
Antibacterial activity of metal-phenanthroline complexes against multidrug-resistant Irish clinical isolates: a whole genome sequencing approach. J Biol Inorg Chem 2023; 28:153-171. [PMID: 36484826 PMCID: PMC9734640 DOI: 10.1007/s00775-022-01979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/08/2022] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance (AMR) is one of the serious global health challenges of our time. There is now an urgent need to develop novel therapeutic agents that can overcome AMR, preferably through alternative mechanistic pathways from conventional treatments. The antibacterial activity of metal complexes (metal = Cu(II), Mn(II), and Ag(I)) incorporating 1,10-phenanthroline (phen) and various dianionic dicarboxylate ligands, along with their simple metal salt and dicarboxylic acid precursors, against common AMR pathogens were investigated. Overall, the highest level of antibacterial activity was evident in compounds that incorporate the phen ligand compared to the activities of their simple salt and dicarboxylic acid precursors. The chelates incorporating both phen and the dianion of 3,6,9-trioxaundecanedioic acid (tdda) were the most effective, and the activity varied depending on the metal centre. Whole-genome sequencing (WGS) was carried out on the reference Pseudomonas aeruginosa strain, PAO1. This strain was exposed to sub-lethal doses of lead metal-tdda-phen complexes to form mutants with induced resistance properties with the aim of elucidating their mechanism of action. Various mutations were detected in the mutant P. aeruginosa genome, causing amino acid changes to proteins involved in cellular respiration, the polyamine biosynthetic pathway, and virulence mechanisms. This study provides insights into acquired resistance mechanisms of pathogenic organisms exposed to Cu(II), Mn(II), and Ag(I) complexes incorporating phen with tdda and warrants further development of these potential complexes as alternative clinical therapeutic drugs to treat AMR infections.
Collapse
|
35
|
Yan X, Zhou Y, Li H, Jiang G, Sun H. Metallomics and metalloproteomics. COMPREHENSIVE INORGANIC CHEMISTRY III 2023:53-76. [DOI: 10.1016/b978-0-12-823144-9.00060-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
36
|
Xiang QQ, Kang YH, Lian LH, Chen ZY, Wang P, Hu JM, Chen LQ. Proteomic profiling reveals mitochondrial toxicity of nanosilver and silver nitrate in the gill of common carp. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 252:106318. [PMID: 36206702 DOI: 10.1016/j.aquatox.2022.106318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
Mitochondria are recognized as an important target organelle for the toxicity of nanomaterials. Although the toxic effects of silver nanoparticles (AgNPs) on mitochondria have been widely reported, the mechanism behind the toxicity remains unclear. In this study, the effects of two forms of silver (AgNPs and AgNO3) on carp gill mitochondria were investigated by analyzing the mitochondrial ultrastructure, physicochemical properties of mitochondrial membrane, and mitochondrial proteomics. After exposure of common carp to AgNPs (0.75 mg/L) and AgNO3 (0.05 mg/L) for 96 h, both forms of silver were shown to cause gill mitochondrial lesions, including irregular shape, loss of mitochondrial cristae, and increased mitochondrial membrane permeability. Proteomics results revealed that AgNPs and AgNO3 induced 362 and 297 differentially expressed proteins (DEPs) in gill mitochondria, respectively. Among the DEPs, 244 were shared between AgNPs and AgNO3 treatments. These shared proteins were mainly distributed in the mitochondrial membrane and matrix, and were significantly enriched in the tricarboxylic acid (TCA) cycle and oxidative phosphorylation pathway. The functional annotation of DEPs induced by both silver forms was mainly involved in energy production and conversion. These results indicated that the toxic mechanism of AgNPs and AgNO3 on gill mitochondria were comparable and the two forms of silver caused mitochondrial dysfunction in fish gills by inhibiting the TCA cycle and disrupting the electron transport chain.
Collapse
Affiliation(s)
- Qian-Qian Xiang
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China
| | - Yu-Hang Kang
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China
| | - Li-Hong Lian
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China
| | - Zhi-Ying Chen
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China
| | - Peng Wang
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; School of Ecology and Environmental Sciences, Yunnan University, Kunming 650091, PR China
| | - Jin-Ming Hu
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China
| | - Li-Qiang Chen
- Instititue of International Rivers and Eco-security, Yunnan Key Laboratory of International Rivers and Trans-Boundary Eco-security, Yunnan University, Kunming 650091, PR China; Yunnan International Joint Research Center for Hydro-Ecology Science & Engineering, Yunnan University, Kunming 650091, PR China.
| |
Collapse
|
37
|
Krishnani KK, Boddu VM, Chadha NK, Chakraborty P, Kumar J, Krishna G, Pathak H. Metallic and non-metallic nanoparticles from plant, animal, and fisheries wastes: potential and valorization for application in agriculture. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:81130-81165. [PMID: 36203045 PMCID: PMC9540199 DOI: 10.1007/s11356-022-23301-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/23/2022] [Indexed: 05/06/2023]
Abstract
Global agriculture is facing tremendous challenges due to climate change. The most predominant amongst these challenges are abiotic and biotic stresses caused by increased incidences of temperature extremes, drought, unseasonal flooding, and pathogens. These threats, mostly due to anthropogenic activities, resulted in severe challenges to crop and livestock production leading to substantial economic losses. It is essential to develop environmentally viable and cost-effective green processes to alleviate these stresses in the crops, livestock, and fisheries. The application of nanomaterials in farming practice to minimize nutrient losses, pest management, and enhance stress resistance capacity is of supreme importance. This paper explores innovative methods for synthesizing metallic and non-metallic nanoparticles using plants, animals, and fisheries wastes and their valorization to mitigate abiotic and biotic stresses and input use efficiency in climate-smart and stress-resilient agriculture including crop plants, livestock, and fisheries.
Collapse
Affiliation(s)
- Kishore Kumar Krishnani
- ICAR-Central Institute of Fisheries Education (Deemed University), Mumbai 400061, Versova, Andheri (W), India.
| | - Veera Mallu Boddu
- Center for Environmental Solutions & Emergency Response (CESER), U.S. Environmental Protection Agency, Research Triangle Park, Durham, NC, USA
| | - Narinder Kumar Chadha
- ICAR-Central Institute of Fisheries Education (Deemed University), Mumbai 400061, Versova, Andheri (W), India
| | - Puja Chakraborty
- ICAR-Central Institute of Fisheries Education (Deemed University), Mumbai 400061, Versova, Andheri (W), India
| | - Jitendra Kumar
- Institute of Pesticide Formulation Technology, Gurugram, Haryana, India
| | - Gopal Krishna
- ICAR-Central Institute of Fisheries Education (Deemed University), Mumbai 400061, Versova, Andheri (W), India
| | - Himanshu Pathak
- Indian Council of Agricultural Research, Krishi Bhavan, New Delhi, 110012, India
| |
Collapse
|
38
|
Frei A, Elliott AG, Kan A, Dinh H, Bräse S, Bruce AE, Bruce MR, Chen F, Humaidy D, Jung N, King AP, Lye PG, Maliszewska HK, Mansour AM, Matiadis D, Muñoz MP, Pai TY, Pokhrel S, Sadler PJ, Sagnou M, Taylor M, Wilson JJ, Woods D, Zuegg J, Meyer W, Cain AK, Cooper MA, Blaskovich MAT. Metal Complexes as Antifungals? From a Crowd-Sourced Compound Library to the First In Vivo Experiments. JACS AU 2022; 2:2277-2294. [PMID: 36311838 PMCID: PMC9597602 DOI: 10.1021/jacsau.2c00308] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
There are currently fewer than 10 antifungal drugs in clinical development, but new fungal strains that are resistant to most current antifungals are spreading rapidly across the world. To prevent a second resistance crisis, new classes of antifungal drugs are urgently needed. Metal complexes have proven to be promising candidates for novel antibiotics, but so far, few compounds have been explored for their potential application as antifungal agents. In this work, we report the evaluation of 1039 metal-containing compounds that were screened by the Community for Open Antimicrobial Drug Discovery (CO-ADD). We show that 20.9% of all metal compounds tested have antimicrobial activity against two representative Candida and Cryptococcus strains compared with only 1.1% of the >300,000 purely organic molecules tested through CO-ADD. We identified 90 metal compounds (8.7%) that show antifungal activity while not displaying any cytotoxicity against mammalian cell lines or hemolytic properties at similar concentrations. The structures of 21 metal complexes that display high antifungal activity (MIC ≤1.25 μM) are discussed and evaluated further against a broad panel of yeasts. Most of these have not been previously tested for antifungal activity. Eleven of these metal complexes were tested for toxicity in the Galleria mellonella moth larva model, revealing that only one compound showed signs of toxicity at the highest injected concentration. Lastly, we demonstrated that the organo-Pt(II) cyclooctadiene complex Pt1 significantly reduces fungal load in an in vivo G. mellonella infection model. These findings showcase that the structural and chemical diversity of metal-based compounds can be an invaluable tool in the development of new drugs against infectious diseases.
Collapse
Affiliation(s)
- Angelo Frei
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
- Department
of Chemistry, Biochemistry & Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012Bern, Switzerland
| | - Alysha G. Elliott
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Alex Kan
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Hue Dinh
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Stefan Bräse
- Institute
of Organic Chemistry, Karlsruhe Institute
of Technology, Fritz-Haber-Weg 6, 76131Karlsruhe, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - Alice E. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Mitchell R. Bruce
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Feng Chen
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Dhirgam Humaidy
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Nicole Jung
- Karlsruhe
Nano Micro Facility (KNMF), Karlsruhe Institute
of Technology, Hermann-von-Helmholtz-Platz 1, 76344Eggenstein-Leopoldshafen, Germany
- Institute
of Biological and Chemical Systems - Functional Molecular Systems, Karlsruhe Institute of Technology, 76344Eggenstein-Leopoldshafen, Germany
| | - A. Paden King
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Peter G. Lye
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Hanna K. Maliszewska
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Ahmed M. Mansour
- Chemistry
Department, Faculty of Science, Cairo University, Giza12613, Egypt
| | - Dimitris Matiadis
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - María Paz Muñoz
- School
of Chemistry, University of East Anglia, Norwich Research Park, NorwichNR4 7TJ, U.K.
| | - Tsung-Yu Pai
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Shyam Pokhrel
- Department
of Chemistry, University of Maine, Orono, Maine04469, United States
| | - Peter J. Sadler
- Department
of Chemistry, University of Warwick, Gibbet Hill Road, CoventryCV4 7AL, U.K.
| | - Marina Sagnou
- Institute
of Biosciences & Applications, National
Centre for Scientific Research “Demokritos”, 15310Athens, Greece
| | - Michelle Taylor
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Justin J. Wilson
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York14853, United States
| | - Dean Woods
- School
of Science and Technology, University of
New England, Armidale, NSW2351, Australia
| | - Johannes Zuegg
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Wieland Meyer
- Molecular
Mycology Research Laboratory, Centre for Infectious Diseases and Microbiology,
Faculty of Medicine and Health, Sydney Medical School, Westmead Clinical
School, Sydney Institute for Infectious Diseases, Westmead Hospital-Research
and Education Network, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW2145, Australia
| | - Amy K. Cain
- School
of Natural Sciences, ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW2109, Australia
| | - Matthew A. Cooper
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland4072, Australia
| |
Collapse
|
39
|
Alahmad A, Al-Zereini WA, Hijazin TJ, Al-Madanat OY, Alghoraibi I, Al-Qaralleh O, Al-Qaraleh S, Feldhoff A, Walter JG, Scheper T. Green Synthesis of Silver Nanoparticles Using Hypericum perforatum L. Aqueous Extract with the Evaluation of Its Antibacterial Activity against Clinical and Food Pathogens. Pharmaceutics 2022; 14:pharmaceutics14051104. [PMID: 35631691 PMCID: PMC9144328 DOI: 10.3390/pharmaceutics14051104] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
The rapid development of nanotechnology and its applications in medicine has provided the perfect solution against a wide range of different microbes, especially antibiotic-resistant ones. In this study, a one-step approach was used in preparing silver nanoparticles (AgNPs) by mixing silver nitrate with hot Hypericum perforatum (St. John’s wort) aqueous extract under high stirring to prevent agglomeration. The formation of silver nanoparticles was monitored by continuous measurement of the surface plasma resonance spectra (UV-VIS). The effect of St. John’s wort aqueous extract on the formation of silver nanoparticles was evaluated and fully characterized by using different physicochemical techniques. The obtained silver nanoparticles were spherical, monodisperse, face-centered cubic (fcc) crystal structures, and the size ranges between 20 to 40 nm. They were covered with a capping layer of organic compounds considered as a nano dimension protective layer that prevents agglomeration and sedimentation. AgNPs revealed antibacterial activity against both tested Gram-positive and Gram-negative bacterial strains causing the formation of 13–32 mm inhibition zones with MIC 6.25–12.5 µg/mL; Escherichia coli strains were resistant to tested AgNPs. The specific growth rate of S. aureus was significantly reduced due to tested AgNPs at concentrations ≥½ MIC. AgNPs did not affect wound migration in fibroblast cell lines compared to control. Our results highlighted the potential use of AgNPs capped with plant extracts in the pharmaceutical and food industries to control bacterial pathogens’ growth; however, further studies are required to confirm their wound healing capability and their health impact must be critically evaluated.
Collapse
Affiliation(s)
- Abdalrahim Alahmad
- Institut für Technische Chemie, Leibniz Universität Hannove, Callinstraße 5, 30167 Hannover, Germany; (J.-G.W.); (T.S.)
- Correspondence: or (A.A.); (W.A.A.-Z.); (O.Y.A.-M.); Tel.: +49-511-7622773 (A.A.); +962-3-2372380 (W.A.A.-Z. & O.Y.A.-M.)
| | - Wael A. Al-Zereini
- Department of Biological Sciences, Faculty of Scince, Mutah University, P.O. Box 7, Mutah 61710, Jordan; (T.J.H.); (O.A.-Q.)
- Correspondence: or (A.A.); (W.A.A.-Z.); (O.Y.A.-M.); Tel.: +49-511-7622773 (A.A.); +962-3-2372380 (W.A.A.-Z. & O.Y.A.-M.)
| | - Tahani J. Hijazin
- Department of Biological Sciences, Faculty of Scince, Mutah University, P.O. Box 7, Mutah 61710, Jordan; (T.J.H.); (O.A.-Q.)
| | - Osama Y. Al-Madanat
- Department of Chemistry, Faculty of Scince, Mutah University, P.O. Box 7, Mutah 61710, Jordan
- Correspondence: or (A.A.); (W.A.A.-Z.); (O.Y.A.-M.); Tel.: +49-511-7622773 (A.A.); +962-3-2372380 (W.A.A.-Z. & O.Y.A.-M.)
| | - Ibrahim Alghoraibi
- Physics Department, Faculty of Science, Damascus University, Damascus P.O. Box 30621, Syria;
| | - Omar Al-Qaralleh
- Department of Biological Sciences, Faculty of Scince, Mutah University, P.O. Box 7, Mutah 61710, Jordan; (T.J.H.); (O.A.-Q.)
| | - Samer Al-Qaraleh
- Faculty of Medicine, Mutah University, P.O. Box 7, Mutah 61710, Jordan;
| | - Armin Feldhoff
- Institut für Physikalische Chemie und Elektrochemie, Leibniz Universität Hannove, Callinstraße 3A, 30167 Hannover, Germany;
| | - Johanna-Gabriela Walter
- Institut für Technische Chemie, Leibniz Universität Hannove, Callinstraße 5, 30167 Hannover, Germany; (J.-G.W.); (T.S.)
| | - Thomas Scheper
- Institut für Technische Chemie, Leibniz Universität Hannove, Callinstraße 5, 30167 Hannover, Germany; (J.-G.W.); (T.S.)
| |
Collapse
|
40
|
Toczek J, Sadłocha M, Major K, Stojko R. Benefit of Silver and Gold Nanoparticles in Wound Healing Process after Endometrial Cancer Protocol. Biomedicines 2022; 10:679. [PMID: 35327481 PMCID: PMC8945154 DOI: 10.3390/biomedicines10030679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/08/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
It is intractable to manage the vast majority of wounds in a classical surgical manner, however if silver, likewise gold and its representative nanoparticles, can lead to the amelioration of the wound healing process after extensive procedures, they should be employed in the current gynecological practice as promptly as possible. Most likely due to its antimicrobial properties, silver is usually applied as an additional component in the wound healing process. In wound management, we obtained various aspects that can lead to impaired wound healing; the crucial aspect for the wound milieu is to prevent the offending agents from occurring. The greatest barrier to healing is represented by the bacterial biofilm, which can occur naturally or in other ways. Biofilm bacteria can produce extracellular polymers, which can then resist concentrated anti-bacterial treatment. The published literature on the use of silver nanoparticles' utilization in wound healing becomes slightly heterogenous and requires us in difficult moments to set up proper treatment guidelines.
Collapse
Affiliation(s)
- Jakub Toczek
- Department of Gynecology, Obstetrics and Oncological Gynecology, Medical University of Silesia in Katowice, Markiefki 87, 40-211 Katowice, Poland; (M.S.); (K.M.); (R.S.)
| | | | | | | |
Collapse
|
41
|
Re-sensitization of mcr carrying multidrug resistant bacteria to colistin by silver. Proc Natl Acad Sci U S A 2022; 119:e2119417119. [PMID: 35263219 PMCID: PMC8931383 DOI: 10.1073/pnas.2119417119] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Superbugs carrying a mobile colistin resistance gene (mcr) are jeopardizing the clinical efficacy of the last-line antibiotic colistin. The development of MCR inhibitors is urgently required to cope with antibiotic-resistance emergencies. Here, we show that silver (Ag+) fully restores the susceptibility of mcr-1–carrying superbugs against colistin both in vitro and in vivo. We found an unprecedented tetra-silver center in the active-site pocket of MCR-1 through the substitution of the essential Zn2+ ions in the intact enzyme, leading to the prevention of substrate binding (i.e. the dysfunction of MCR-1 in transferring phosphorylethanolamine to lipid A). Importantly, the ability of Ag+ to suppress resistance evolution extends the lifespan of currently used antibiotics, providing a strategy to treat infections by mcr-positive bacteria. Colistin is considered the last-line antimicrobial for the treatment of multidrug-resistant gram-negative bacterial infections. The emergence and spread of superbugs carrying the mobile colistin resistance gene (mcr) have become the most serious and urgent threat to healthcare. Here, we discover that silver (Ag+), including silver nanoparticles, could restore colistin efficacy against mcr-positive bacteria. We show that Ag+ inhibits the activity of the MCR-1 enzyme via substitution of Zn2+ in the active site. Unexpectedly, a tetra-silver center was found in the active-site pocket of MCR-1 as revealed by the X-ray structure of the Ag-bound MCR-1, resulting in the prevention of substrate binding. Moreover, Ag+ effectively slows down the development of higher-level resistance and reduces mutation frequency. Importantly, the combined use of Ag+ at a low concentration with colistin could relieve dermonecrotic lesions and reduce the bacterial load of mice infected with mcr-1–carrying pathogens. This study depicts a mechanism of Ag+ inhibition of MCR enzymes and demonstrates the potentials of Ag+ as broad-spectrum inhibitors for the treatment of mcr-positive bacterial infection in combination with colistin.
Collapse
|
42
|
Kluska K, Veronesi G, Deniaud A, Hajdu B, Gyurcsik B, Bal W, Krężel A. Structures of Silver Fingers and a Pathway to Their Genotoxicity. Angew Chem Int Ed Engl 2022; 61:e202116621. [PMID: 35041243 PMCID: PMC9303758 DOI: 10.1002/anie.202116621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Recently, we demonstrated that AgI can directly replace ZnII in zinc fingers (ZFs). The cooperative binding of AgI to ZFs leads to a thermodynamically irreversible formation of silver clusters destroying the native ZF structure. Thus, a reported loss of biological function of ZF proteins is a likely consequence of such replacement. Here, we report an X-ray absorption spectroscopy (XAS) study of Agn Sn clusters formed in ZFs to probe their structural features. Selective probing of the local environment around AgI by XAS showed the predominance of digonal AgI coordination to two sulfur donors, coordinated with an average Ag-S distance at 2.41 Å. No Ag-N bonds were present. A mixed AgS2 /AgS3 geometry was found solely in the CCCH AgI -ZF. We also show that cooperative replacement of ZnII ions with the studied Ag2 S2 clusters occurred in a three-ZF transcription factor protein 1MEY#, leading to a dissociation of 1MEY# from the complex with its cognate DNA.
Collapse
Affiliation(s)
- Katarzyna Kluska
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, 50-383, Wrocław, Poland
| | - Giulia Veronesi
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, 38000, Grenoble, France
| | - Aurélien Deniaud
- Univ. Grenoble Alpes, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, 38000, Grenoble, France
| | - Bálint Hajdu
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, 6720, Szeged, Hungary
| | - Béla Gyurcsik
- Department of Inorganic and Analytical Chemistry, Faculty of Science and Informatics, University of Szeged, Dóm tér 7, 6720, Szeged, Hungary
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawińskiego 5a, 02-106, Warsaw, Poland
| | - Artur Krężel
- Department of Chemical Biology, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a, 50-383, Wrocław, Poland
| |
Collapse
|
43
|
Abstract
Metals are essential components in life processes and participate in many important biological processes. Dysregulation of metal homeostasis is correlated with many diseases. Metals are also frequently incorporated into diagnosis and therapeutics. Understanding of metal homeostasis under (patho)physiological conditions and the molecular mechanisms of action of metallodrugs in biological systems has positive impacts on human health. As an emerging interdisciplinary area of research, metalloproteomics involves investigating metal-protein interactions in biological systems at a proteome-wide scale, has received growing attention, and has been implemented into metal-related research. In this review, we summarize the recent advances in metalloproteomics methodologies and applications. We also highlight emerging single-cell metalloproteomics, including time-resolved inductively coupled plasma mass spectrometry, mass cytometry, and secondary ion mass spectrometry. Finally, we discuss future perspectives in metalloproteomics, aiming to attract more original research to develop more advanced methodologies, which could be utilized rapidly by biochemists or biologists to expand our knowledge of how metal functions in biology and medicine. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| | - Hongyan Li
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| | - Hongzhe Sun
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics on Health and Environment, University of Hong Kong, Hong Kong SAR, China; ,
| |
Collapse
|
44
|
Abstract
Antibiotic resistance has caused a serious threat to public health and human safety. Recently, the emergence of novel resistance gene tet(X4) and its variants threatens the clinical utility of tigecycline, one of the last-line antibiotics for multidrug-resistant (MDR) bacterial infections. It is highly promising to develop effective antibiotic adjuvants to restore the clinical efficacy of existing drugs and extend their life spans. Metal compounds, such as silver, have been widely used as potential antimicrobial agents for decades. However, the potentiating effect of metallo-agents on the existing antibiotics is not fully understood. Here, we found that five bismuth drugs, especially bismuth nitrate [Bi(NO3)3], commonly used in clinical treatment of stomach-associated diseases, effectively boost the antibacterial activity of tigecycline against tet(X)-positive bacteria by inhibiting the enzymatic activity of Tet(X) protein. Furthermore, the combination of Bi(NO3)3 and tigecycline prevents the development of higher-level resistance in Tet(X)-expressing Gram-negative bacteria. Using molecular docking and dynamics simulation assays, we revealed that Bi(NO3)3 can competitively bind to the active center of Tet(X4) protein, while the bismuth atom targets the Tet(X4) protein in a noncompetitive manner and changes the structure of the primary binding pocket. These two mechanisms of action both antagonize the enzymatic activity of Tet(X4) resistance protein on tigecycline. Collectively, these findings indicate the high potential of bismuth drugs as novel Tet(X) inhibitors to treat tet(X4)-positive bacteria-associated infections in combination with tigecycline. IMPORTANCE Recently, high-level tigecycline resistance mediated by tet(X4) and its variants represents a serious challenge for global public health. Antibiotic adjuvant strategy that enhances the activity of the existing antibiotics by using nonantibiotic drugs offers a distinct approach to combat the antibiotic resistance crisis. In this study, we found that bismuth drugs involve bismuth nitrate, a compound previously approved for treatment of stomach-associated diseases, remarkably potentiates tigecycline activity against tet(X)-positive bacteria. Mechanistic studies showed that bismuth drugs effectively suppress the enzymatic activity of Tet(X) resistance protein. Specifically, bismuth nitrate targets the active center of Tet(X4) protein, while bismuth binds to the resistance protein in a noncompetitive manner. Our data open up a new horizon for the treatment of infections caused by tet(X)-bearing superbugs.
Collapse
|
45
|
Kluska K, Veronesi G, Deniaud A, Hajdu B, Gyurcsik B, Bal W, Krezel A. Structures of Silver Fingers and a Pathway to Their Genotoxicity. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202116621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Katarzyna Kluska
- University of Wroclaw: Uniwersytet Wroclawski Faculty of Biotechnology, Department of Chemical Biology 50-383 Wrocław POLAND
| | - Giulia Veronesi
- Université Grenoble Alpes: Universite Grenoble Alpes Laboratoire de Chimie et Biologie des Metaux F-38000 Grenoble FRANCE
| | - Aurelien Deniaud
- Université de Grenoble I: Universite Grenoble Alpes Laboratoire de Chimie at Biologie des Metaux F-38000 Grenoble FRANCE
| | - Balint Hajdu
- University of Szeged: Szegedi Tudomanyegyetem Department of Inorganic Analytical Chemistry H-6720 Szeged HUNGARY
| | - Bela Gyurcsik
- University of Szeged: Szegedi Tudomanyegyetem Depertment of Inorganic Analytical Chemistry H-6720 Szeged HUNGARY
| | - Wojciech Bal
- Polish Academy of Sciences: Polska Akademia Nauk Institute of Biochemistry and Biophysics 02-106 Warsaw POLAND
| | - Artur Krezel
- University of Wroclaw Department of Chemical Biology, Faculty of Biotechnology F. Joliot-Curie 14A 50-383 Wrocław POLAND
| |
Collapse
|
46
|
Wilkinson HN, Guinn BA, Hardman MJ. Combined Metallomics/Transcriptomics Profiling Reveals a Major Role for Metals in Wound Repair. Front Cell Dev Biol 2021; 9:788596. [PMID: 34917621 PMCID: PMC8669724 DOI: 10.3389/fcell.2021.788596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Endogenous metals are required for all life, orchestrating the action of diverse cellular processes that are crucial for tissue function. The dynamic wound healing response is underpinned by a plethora of such cellular behaviours, occurring in a time-dependent manner. However, the importance of endogenous metals for cutaneous repair remains largely unexplored. Here we combine ICP-MS with tissue-level RNA-sequencing to reveal profound changes in a number of metals, and corresponding metal-regulated genes, across temporal healing in mice. Wound calcium, magnesium, iron, copper and manganese are elevated at 7 days post-wounding, while magnesium, iron, aluminium, manganese and cobalt increase at 14 days post-wounding. At the level of transcription, wound-induced pathways are independently highly enriched for metal-regulated genes, and vice versa. Moreover, specific metals are linked to distinct wound-induced biological processes and converge on key transcriptional regulators in mice and humans. Finally, we reveal a potential role for one newly identified transcriptional regulator, TNF, in calcium-induced epidermal differentiation. Together, these data highlight potential new and diverse roles for metals in cutaneous wound repair, paving the way for further studies to elucidate the contribution of metals to cellular processes in the repair of skin and other tissues.
Collapse
Affiliation(s)
- Holly N Wilkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, Hull, United Kingdom
| | - Barbara-Ann Guinn
- Department of Biomedical Sciences, Faculty of Health, The University of Hull, Hull, United Kingdom
| | - Matthew J Hardman
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, The University of Hull, Hull, United Kingdom
| |
Collapse
|
47
|
Long-term, synergistic and high-efficient antibacterial polyacrylonitrile nanofibrous membrane prepared by "one-pot" electrospinning process. J Colloid Interface Sci 2021; 609:718-733. [PMID: 34863546 DOI: 10.1016/j.jcis.2021.11.075] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 01/07/2023]
Abstract
Enhancing long-term antibacterial activity of membrane materials is an effective strategy to reduce biological contamination. Herein, we developed a long-term, synergistic antibacterial polyacrylonitrile (PAN) nanofiber membrane by a "one-pot" electrospinning process. In the reaction solution of PAN and N, N-dimethylformamide (DMF), silver-silicon dioxide nanoparticles (Ag@SiO2 NPs) are in-situ synthesized and stabilized using silane coupling agent; and [2-(methacryloyloxy)-ethyl] trimethylammonium chloride (MT) monomers are then in-situ cross-linked to obtain a polyquaternary ammonium salt (PMT). Subsequently, the casting solution is directly used to fabricate Ag@SiO2/PMT-PAN nanofibrous membrane (NFM) via electrospinning. The antibacterial activity, reusability, synergy effect and biological safety of the Ag@SiO2/PMT-PAN NFM are systematically investigated, and the synergistic antibacterial mechanism is also explored. Even at very low (0.3 wt%) content of silver, the Ag@SiO2/PMT-PAN NFM exhibits excellent antibacterial activity against E. coli (99%) and S. aureus (99%). Also, the antibacterial ability of the NFM remains the same level after three cycles of antibacterial processes with the efficient synergy effects of Ag@SiO2 and PMT components. When the Ag@SiO2/PMT-PAN contacts with bacteria, the PMT attracts and kills the bacteria through electrostatic action. The bacteria with damaged cell membranes are deposited on the nanofibrous membrane, which could greatly promote the release of Ag+ and further enhance the antibacterial activity. Moreover, L929 fibroblasts are co-cultured with the extract of 4 mg/mL Ag@SiO2/PMT-PAN for 5 days, which exhibits a low cytotoxicity with a cell proliferation ratio of 95%. This work opens new pathways for developing long-term effective and synergistic antibacterial nanofibrous membrane materials to prevent infections associated with biomedical equipment.
Collapse
|
48
|
Griffith DM, Li H, Werrett MV, Andrews PC, Sun H. Medicinal chemistry and biomedical applications of bismuth-based compounds and nanoparticles. Chem Soc Rev 2021; 50:12037-12069. [PMID: 34533144 DOI: 10.1039/d0cs00031k] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bismuth as a relatively non-toxic and inexpensive metal with exceptional properties has numerous biomedical applications. Bismuth-based compounds are used extensively as medicines for the treatment of gastrointestinal disorders including dyspepsia, gastric ulcers and H. pylori infections. Recently, its medicinal application was further extended to potential treatments of viral infection, multidrug resistant microbial infections, cancer and also imaging, drug delivery and biosensing. In this review we have highlighted the unique chemistry and biological chemistry of bismuth-209 as a prelude to sections covering the unique antibacterial activity of bismuth including a description of research undertaken to date to elucidate key molecular mechanisms of action against H. pylori, the development of novel compounds to treat infection from microbes beyond H. pylori and the significant role bismuth compounds can play as resistance breakers. Furthermore we have provided an account of the potential therapeutic application of bismuth-213 in targeted alpha therapy as well as a summary of the biomedical applications of bismuth-based nanoparticles and composites. Ultimately this review aims to provide the state of the art, highlight the untapped biomedical potential of bismuth and encourage original contributions to this exciting and important field.
Collapse
Affiliation(s)
- Darren M Griffith
- Department of Chemistry, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland.,SSPC, Synthesis and Solid State Pharmaceutical Centre, Ireland
| | - Hongyan Li
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| | | | - Philip C Andrews
- School of Chemistry, Monash University, Melbourne, VIC, Australia
| | - Hongzhe Sun
- Department of Chemistry and CAS-HKU Joint Laboratory of Metallomics for Health and Environment, The University of Hong Kong, Pokfulam Road, Hong Kong, China.
| |
Collapse
|
49
|
Betts HD, Neville SL, McDevitt CA, Sumby CJ, Harris HH. The biochemical fate of Ag + ions in Staphylococcus aureus, Escherichia coli, and biological media. J Inorg Biochem 2021; 225:111598. [PMID: 34517168 DOI: 10.1016/j.jinorgbio.2021.111598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/03/2021] [Accepted: 08/28/2021] [Indexed: 01/16/2023]
Abstract
Silver is commonly included in a range of household and medical items to provide bactericidal action. Despite this, the chemical fate of the metal in both mammalian and bacterial systems remains poorly understood. Here, we applied a metallomics approach using X-ray absorption spectroscopy (XAS) and size-exclusion chromatography hyphenated with inductively coupled plasma mass spectrometry (SEC-ICP-MS) to advance our understanding of the biochemical fate of silver ions in bacterial culture and cells, and the chemistry associated with these interactions. When silver ions were added to lysogeny broth, silver was exclusively associated with moderately-sized species (~30 kDa) and bound by thiolate ligands. In two representative bacterial pathogens cultured in lysogeny broth including sub-lethal concentrations of ionic silver, silver was found in cells to be predominantly coordinated by thiolate species. The silver biomacromolecule-binding profile in Staphylococcus aureus and Escherichia coli was complex, with silver bound by a range of species spanning from 20 kDa to >1220 kDa. In bacterial cells, silver was nonuniformly colocalised with copper-bound proteins, suggesting that cellular copper processing may, in part, confuse silver for nutrient copper. Notably, in the treated cells, silver was not detected bound to low molecular weight compounds such as glutathione or bacillithiol.
Collapse
Affiliation(s)
- Harley D Betts
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - Stephanie L Neville
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Christopher A McDevitt
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria 3000, Australia
| | - Christopher J Sumby
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia
| | - Hugh H Harris
- Department of Chemistry, The University of Adelaide, South Australia 5005, Australia,.
| |
Collapse
|
50
|
Multi-target mode of action of silver against Staphylococcus aureus endows it with capability to combat antibiotic resistance. Nat Commun 2021; 12:3331. [PMID: 34099682 PMCID: PMC8184742 DOI: 10.1038/s41467-021-23659-y] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
The rapid emergence of drug resistant Staphylococcus aureus (S. aureus) poses a serious threat to public health globally. Silver (Ag)-based antimicrobials are promising to combat antibiotic resistant S. aureus, yet their molecular targets are largely elusive. Herein, we separate and identify 38 authentic Ag+-binding proteins in S. aureus at the whole-cell scale. We then capture the molecular snapshot on the dynamic action of Ag+ against S. aureus and further validate that Ag+ could inhibit a key target 6-phosphogluconate dehydrogenase through binding to catalytic His185 by X-ray crystallography. Significantly, the multi-target mode of action of Ag+ (and nanosilver) endows its sustainable antimicrobial efficacy, leading to enhanced efficacy of conventional antibiotics and resensitization of MRSA to antibiotics. Our study resolves the long-standing question of the molecular targets of silver in S. aureus and offers insights into the sustainable bacterial susceptibility of silver, providing a potential approach for combating antimicrobial resistance. Silver (Ag) has been used as an antimicrobial agent since a long time, but its molecular mechanism of action was not elucidated due to technical challenges. Here, the authors develop a mass spectrometric approach to identify the Ag-proteome in Staphylococcus aureus, and capture a molecular snapshot of the dynamic bactericidal mode of action of Ag through targeting multiple biological pathways.
Collapse
|